Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 107(3): 473-486, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32781046

RESUMEN

Africa contains more human genetic variation than any other continent, but the majority of the population-scale analyses of the African peoples have focused on just two of the four major linguistic groups, the Niger-Congo and Afro-Asiatic, leaving the Nilo-Saharan and Khoisan populations under-represented. In order to assess genetic variation and signatures of selection within a Nilo-Saharan population and between the Nilo-Saharan and Niger-Congo and Afro-Asiatic, we sequenced 50 genomes from the Nilo-Saharan Lugbara population of North-West Uganda and 250 genomes from 6 previously unsequenced Niger-Congo populations. We compared these data to data from a further 16 Eurasian and African populations including the Gumuz, another putative Nilo-Saharan population from Ethiopia. Of the 21 million variants identified in the Nilo-Saharan population, 3.57 million (17%) were not represented in dbSNP and included predicted non-synonymous mutations with possible phenotypic effects. We found greater genetic differentiation between the Nilo-Saharan Lugbara and Gumuz populations than between any two Afro-Asiatic or Niger-Congo populations. F3 tests showed that Gumuz contributed a genetic component to most Niger-Congo B populations whereas Lugabara did not. We scanned the genomes of the Lugbara for evidence of selective sweeps. We found selective sweeps at four loci (SLC24A5, SNX13, TYRP1, and UVRAG) associated with skin pigmentation, three of which already have been reported to be under selection. These selective sweeps point toward adaptations to the intense UV radiation of the Sahel.


Asunto(s)
Adaptación Fisiológica/genética , Variación Genética/genética , Selección Genética/genética , Pigmentación de la Piel/genética , Antiportadores/genética , Población Negra/genética , Manejo de Datos , Etiopía/epidemiología , Femenino , Genética de Población , Genoma Humano/genética , Haplotipos/genética , Humanos , Masculino , Glicoproteínas de Membrana/genética , Oxidorreductasas/genética , Polimorfismo de Nucleótido Simple/genética , Nexinas de Clasificación/genética , Proteínas Supresoras de Tumor/genética , Uganda/epidemiología
2.
Bull World Health Organ ; 101(8): 522-528, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37529018

RESUMEN

Human African trypanosomiasis is a life-threatening parasitic infection transmitted by the tsetse fly in sub-Saharan Africa. The most common form is caused by Trypanosoma brucei gambiense, with humans as the main reservoir. Diagnosis in the field requires microscopic examination performed by specifically trained personnel. After over two decades of sustained efforts, the incidence of the disease is strongly declining, and some historically endemic countries are no longer detecting cases. The World Health Organization (WHO) has targeted the elimination of transmission of gambiense human African trypanosomiasis by 2030, defined as zero autochthonous cases for at least five consecutive years. Endemic countries reaching this goal must maintain dedicated surveillance to detect re-emergence or re-introduction. With this new agenda, new tools are needed for verification of the absence of transmission. WHO has therefore developed a target product profile calling for development of a method for population-level cross-cutting surveillance of T. b. gambiense transmission. The method needs to be performed in national or sub-national reference laboratories, and to test in parallel numerous samples shipped from remote rural areas. Among other characteristics the product profile specifies: (i) a simple specimen collection procedure; (ii) no cold-chain requirement to transfer specimens to reference laboratories; (iii) high sensitivity and specificity; (iv) high-throughput, substantially automatized; (v) low cost per specimen, when analysed in large batches; and (vi) applicable also in animals.


La trypanosomiase humaine africaine est une infection parasitaire potentiellement mortelle transmise par la mouche tsé-tsé en Afrique subsaharienne. La forme la plus répandue est causée par Trypanosoma brucei gambiense, les humains constituant son principal réservoir. Établir un diagnostic sur le terrain nécessite un examen microscopique réalisé par du personnel formé à cet effet. Après plus de deux décennies d'efforts soutenus, l'incidence de la maladie diminue fortement et quelques pays historiquement endémiques ne découvrent plus aucun cas. L'objectif de l'Organisation mondiale de la Santé (OMS) est d'éliminer la transmission de la trypanosomiase humaine africaine à T. b. gambiense d'ici 2030, ce qui correspond à zéro cas autochtone pendant au moins cinq années consécutives. Les pays endémiques qui atteignent cet objectif doivent maintenir une surveillance spécifique afin de détecter toute réémergence ou réintroduction. Ce nouveau programme doit s'accompagner de nouveaux outils servant à vérifier l'absence de transmission. L'OMS a donc élaboré un profil de produit cible pour le développement d'un procédé de surveillance transversale de la transmission de T. b. gambiense à l'échelle de la population. Ce procédé doit être effectué dans des laboratoires de référence nationaux ou infranationaux et tester simultanément de nombreux échantillons envoyés depuis des régions rurales isolées. Ce profil de produit comporte notamment les caractéristiques suivantes: (i) une procédure simple de collecte d'échantillons; (ii) aucune exigence concernant le respect de la chaîne du froid lors du transfert des échantillons vers les laboratoires de référence; (iii) un niveau élevé de sensibilité et de spécificité; (iv) un haut débit, en grande partie automatisé; (v) de faibles coûts par échantillon lors d'analyses en masse; et enfin, (vi) applicable aux animaux également.


La tripanosomiasis humana africana es una infección parasitaria potencialmente mortal transmitida por la mosca tsetsé en el África Subsahariana. El principal reservorio es el ser humano, y la forma más común está causada por Trypanosoma brucei gambiense. El diagnóstico práctico requiere un examen microscópico a cargo de personal con formación específica. Tras más de dos décadas de esfuerzos sostenidos, la incidencia de la enfermedad está disminuyendo considerablemente, y en algunos países históricamente endémicos ya no se detectan casos. La Organización Mundial de la Salud (OMS) se ha fijado como objetivo la eliminación de la transmisión de la tripanosomiasis africana humana gambiense para 2030, es decir, cero casos autóctonos durante al menos cinco años consecutivos. Los países endémicos que alcancen este objetivo deben mantener una vigilancia permanente para detectar la reaparición o reintroducción de la enfermedad. Con esta agenda nueva, se necesitan herramientas nuevas para verificar la ausencia de transmisión. Por consiguiente, la OMS ha elaborado un perfil de producto objetivo en el que se pide el desarrollo de un método para la vigilancia transversal a nivel de población sobre la transmisión de T. b. gambiense. El método debe realizarse en laboratorios de referencia nacionales o subnacionales y analizar en paralelo numerosas muestras enviadas desde regiones rurales remotas. Entre otras características, el perfil del producto detalla: (i) un procedimiento sencillo de recogida de muestras; (ii) ningún requisito de cadena de frío para transferir las muestras a los laboratorios de referencia; (iii) alta sensibilidad y especificidad; (iv) alto rendimiento, sustancialmente automatizado; (v) bajo coste por muestra, cuando se analizan en grandes lotes; y (vi) aplicable también en animales.


Asunto(s)
Tripanosomiasis Africana , Moscas Tse-Tse , Animales , Humanos , Trypanosoma brucei gambiense , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/parasitología , Moscas Tse-Tse/parasitología , África del Sur del Sahara , Incidencia
3.
Bull World Health Organ ; 101(8): 529-534, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37529024

RESUMEN

Rhodesiense human African trypanosomiasis is a lethal parasitic infection caused by Trypanosoma brucei rhodesiense and transmitted by tsetse flies in eastern and southern Africa. It accounts for around 5% of all cases of human African trypanosomiasis. Currently, there is no simple serological test for rhodesiense human African trypanosomiasis and diagnosis relies on microscopic confirmation of trypanosomes in samples of blood or other tissues. The availability of a simple and accurate diagnostic test would aid the control, surveillance and treatment of the disease. A subcommittee of the World Health Organization's Neglected Tropical Diseases Diagnostics Technical Advisory Group has developed a target product profile for a diagnostic tool to identify T. b. rhodesiense infection. The optimum tool would have a sensitivity and specificity above 99% for detecting T. b. rhodesiense, but be simple enough for use by minimally trained health-care workers in unsophisticated peripheral health facilities or mobile teams in villages. The test should yield a qualitative result that can be easily observed and can be used to determine treatment. An antigen test would be preferable, with blood collected by finger-prick. Ideally, there should be no need for a cold chain, instrumentation or precision liquid handling. The test should be usable between 10 °C and 40 °C and between 10% and 88% relative humidity. Basic training should take under 2 hours and the test should involve fewer than five steps. The unit cost should be less than 1 United States dollar.


La trypanosomiase humaine africaine à T. b. rhodesiense est une infection parasitaire mortelle causée par Trypanosoma brucei rhodesiense et transmise par les mouches tsé-tsé en Afrique orientale et australe. Elle représente environ 5% de l'ensemble des cas de trypanosomiase humaine africaine. À l'heure actuelle, il n'existe aucun test sérologique simple pour l'infection à T. b. rhodesiense et le diagnostic repose sur la confirmation microscopique de la présence de trypanosomes dans des échantillons de sang ou d'autres tissus. Fournir un test de diagnostic simple et précis favoriserait la lutte, la surveillance et la prise en charge de la maladie. Un sous-comité du Groupe consultatif technique sur les produits de diagnostic des maladies tropicales négligées de l'Organisation mondiale de la Santé a donc élaboré un profil de produit cible pour un outil visant à détecter une infection par T. b. rhodesiense. L'outil le plus adapté présenterait un niveau de sensibilité et de spécificité supérieur à 99% pour la détection de T. b. rhodesiense, tout en étant à la portée de professionnels de la santé ayant reçu une formation sommaire, tant dans des structures de santé périphériques basiques qu'au sein d'équipes mobiles dans les villages. Cet outil doit fournir un résultat fiable, facile à interpréter, qui peut servir à établir un traitement. Un test antigénique serait préférable, avec prélèvement de l'échantillon sanguin par le biais d'une piqûre au bout du doigt. Idéalement, l'outil ne doit pas être thermosensible, ni nécessiter un équipement spécifique ou une manipulation de liquides délicate. Le test doit pouvoir être utilisé à une température comprise entre 10 °C et 40 °C, ainsi que dans une humidité relative de 10% à 88%. La formation requise pour son utilisation doit durer moins de deux heures et le test doit être effectué en moins de cinq étapes, Enfin, son coût unitaire doit être inférieur à un dollar américain.


La tripanosomiasis humana africana rhodesiense es una infección letal parasitaria causada por el Trypanosoma brucei rhodesiense, y es transmitida por la mosca tse-tsé en África oriental y meridional. Representa aproximadamente el 5% de todos los casos de tripanosomiasis humana africana. Actualmente, no existe ninguna prueba serológica simple para la tripanosomiasis humana africana rhodesiense, y el diagnóstico se basa en la confirmación microscópica de tripanosomas existentes en muestras de sangre u otros tejidos. Una prueba diagnóstica sencilla y precisa ayudaría a controlar, vigilar y tratar la enfermedad. Un subcomité del Grupo Asesor Técnico de Diagnóstico de Enfermedades Tropicales Desatendidas de la Organización Mundial de la Salud ha creado un perfil de producto objetivo para una herramienta de diagnóstico que permita identificar la infección T. b. rhodesiense. La herramienta óptima tendría una sensibilidad y una especificidad superiores al 99% para detectar la T. b. rhodesiense y, al ser lo suficientemente sencilla, podrían utilizarla trabajadores sanitarios mínimamente formados, en centros sanitarios periféricos no sofisticados, o bien equipos móviles. La prueba debe arrojar un resultado cualitativo de fácil lectura y que pueda utilizarse para determinar el tratamiento. Sería preferible una prueba de antígenos, con sangre extraída mediante punción digital. Idealmente, no debería ser necesaria la cadena de frío, la instrumentación ni la manipulación de líquidos de precisión. La prueba debe poder utilizarse entre 10 °C y 40 °C, con una humedad relativa de entre el 10% y el 88%. La instrucción básica debe llevar menos de 2 horas y la prueba debe incluir menos de cinco pasos. El coste de la unidad debe ser inferior a 1 dólar estadounidense.


Asunto(s)
Trypanosoma brucei rhodesiense , Tripanosomiasis Africana , Animales , Humanos , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/parasitología , África Austral , Sensibilidad y Especificidad , Pruebas Diagnósticas de Rutina
4.
Bull World Health Organ ; 101(8): 541-545, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37529025

RESUMEN

Having caused devastating epidemics during the 20th century, the incidence of life-threatening human African trypanosomiasis has fallen to historically low levels as a result of sustained and coordinated efforts over the past 20 years. Humans are the main reservoir of one of the two pathogenic trypanosome subspecies, Trypanosoma brucei gambiense, found in western and central Africa. The expected advent of a safe and easy-to-use treatment to be given to seropositive but microscopically unconfirmed individuals would lead to further depletion; in the meantime, the presence of T. b. gambiense infection in the community must be monitored to allow the control strategy to be adapted and the elimination status to be assessed. The World Health Organization has therefore developed a target product profile that describes the optimal and minimal characteristics of an individual laboratory-based test to assess T. b. gambiense infection in low-prevalence settings. Development of the target product profile involved the formation of a Neglected Tropical Diseases Diagnostics Technical Advisory Group and a subgroup on human African trypanosomiasis diagnostic innovation needs, and an analysis of the available products and development pipeline. According to the product profile, the test should ideally: (i) require a minimally invasive or non-invasive specimen, collectable at peripheral facilities by minimally trained health workers; (ii) demonstrate good sensitivity and high specificity; (iii) have a stability of samples allowing transfer to reference laboratories preferably without cold chain; (iv) be stable over a wide range of environmental conditions for more than 2 years; and (v) after marketing, be available at low cost for at least 7 years.


Après avoir causé des épidémies dévastatrices au cours du 20e siècle, la trypanosomiase humaine africaine, potentiellement mortelle, a vu son incidence chuter à un niveau historiquement bas grâce aux efforts conjoints et soutenus déployés ces deux dernières décennies. Les humains constituent le principal réservoir de l'une des deux sous-espèces pathogéniques de trypanosome, Trypanosoma brucei gambiense, que l'on retrouve en Afrique occidentale et centrale. L'arrivée d'un traitement sûr et simple d'utilisation, qui serait administré aux individus séropositifs mais sans confirmation microscopique, devrait entraîner une nouvelle diminution; dans l'intervalle, la présence d'une infection à T. b. gambiense au sein de la communauté doit être surveillée afin de pouvoir adapter la stratégie de lutte et évaluer le statut d'élimination. Par conséquent, l'Organisation mondiale de la Santé a élaboré un profil de produit cible qui détaille les caractéristiques minimales et optimales d'un test individuel en laboratoire visant à confirmer l'infection à T. b. gambiense dans les régions à faible prévalence. La mise au point de ce profil a entraîné la formation d'un Groupe consultatif technique sur le diagnostic des maladies tropicales négligées et d'un sous-groupe consacré aux besoins en matière d'innovation diagnostique pour la trypanosomiase humaine africaine, qui a conduit une analyse des produits existants et des projets de développement. Selon le profil de produit, le test devrait idéalement: (i) nécessiter un prélèvement d'échantillon peu ou non invasif, pouvant être effectué dans des structures périphériques par des professionnels de la santé ayant reçu une formation sommaire; (ii) faire preuve d'un bon niveau de sensibilité et d'un niveau élevé de spécificité; (iii) avoir une stabilité des échantillons permettant le transfert vers des laboratoires de référence, de préférence sans chaîne de froid; (iv) rester stable dans un large éventail de conditions environnementales pendant plus de deux ans; et enfin, (v) après commercialisation, être disponible à bas coût pendant au moins sept ans.


Tras haber causado epidemias devastadoras durante el siglo XX, la incidencia de la tripanosomiasis humana africana potencialmente mortal ha descendido a niveles históricamente bajos gracias a los esfuerzos sostenidos y coordinados de los últimos 20 años. El ser humano es el principal reservorio de una de las dos subespecies patógenas del tripanosoma, Trypanosoma brucei gambiense, presente en África Occidental y Central. La prevista disponibilidad de un tratamiento seguro y fácil de administrar a personas seropositivas, pero no confirmadas al microscopio, permitiría una mayor eliminación; mientras tanto, se debe vigilar la presencia de la infección por T. b. gambiense en la comunidad para poder adaptar la estrategia de control y evaluar el estado de eliminación. Por consiguiente, la Organización Mundial de la Salud ha elaborado un perfil de producto objetivo que describe las características óptimas y mínimas de una prueba de laboratorio individual para evaluar la infección por T. b. gambiense en regiones de baja prevalencia. El desarrollo del perfil de producto objetivo implicó la formación de un Grupo de Asesoramiento Técnico sobre Diagnóstico de Enfermedades Tropicales Desatendidas y un subgrupo sobre las necesidades de innovación en el diagnóstico de la tripanosomiasis humana africana, así como un análisis de los productos disponibles y en desarrollo. Según el perfil objetivo, lo ideal sería que la prueba: (i) requiriera una muestra mínimamente invasiva o no invasiva, que pudiera ser recogida en centros periféricos por personal sanitario con una capacitación mínima; (ii) demostrara una buena sensibilidad y alta especificidad; (iii) tuviera una estabilidad de las muestras que permita su transferencia a laboratorios de referencia, preferiblemente sin cadena de frío; (iv) fuera estable en un amplio rango de condiciones ambientales durante más de 2 años; y (v) tras su comercialización, estuviera disponible a bajo coste durante al menos 7 años.


Asunto(s)
Trypanosoma brucei gambiense , Tripanosomiasis Africana , Animales , Humanos , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/epidemiología , Prevalencia , Incidencia
5.
Bull World Health Organ ; 101(8): 535-540, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37529027

RESUMEN

Human African trypanosomiasis is a life-threatening parasitic infection endemic to sub-Saharan Africa. Around 95% of cases are due to Trypanosoma brucei gambiense, found in western and central Africa. Clinical signs and symptoms are nonspecific, current diagnostic tests are not sufficiently accurate, and parasitological confirmation of infection requires microscopic examination of body fluids and specialized techniques for concentrating parasites. Moreover, current treatment is not recommended on the basis of suspicion alone because it is not sufficiently safe. The availability of a simple and accurate diagnostic test to identify individuals harbouring parasites would widen treatment and help decrease disease prevalence. A subcommittee of the World Health Organization's Neglected Tropical Diseases Diagnostics Technical Advisory Group has developed a target product profile for a diagnostic tool to identify T. b. gambiense infection. This tool should have a high sensitivity for detecting T. b. gambiense but be simple enough to use in rural Africa. Ideally, the tool could be applied by any minimally trained individual in an unsophisticated peripheral health facility, or a mobile team in a village with little infrastructure. The test should be able to function under hot and humid conditions. Basic training should take under 2 hours and the test should involve fewer than five steps. There should be no need for instrumentation or precision liquid handling. The test should yield a qualitative result in under 20 minutes that can be easily observed, and one test should be sufficient for determining treatment. A unit cost below 1 United States dollar (US$) would enable mass screening.


La trypanosomiase humaine africaine est une infection parasitaire potentiellement mortelle endémique en Afrique subsaharienne. Dans près de 95% des cas, elle est causée par Trypanosoma brucei gambiense, que l'on trouve en Afrique occidentale et centrale. Les symptômes et signes cliniques sont aspécifiques, les tests de diagnostic existants ne sont pas assez précis et la confirmation parasitologique de l'infection nécessite un examen microscopique des liquides corporels ainsi que des techniques spécialisées pour concentrer les parasites. En outre, il n'est pas recommandé d'entamer le traitement actuel sur la base d'une simple suspicion car celui-ci n'est pas suffisamment sûr. Fournir un test de diagnostic simple et précis permettant d'identifier les individus porteurs de parasites contribuerait à élargir le traitement et à une diminution de la prévalence de la maladie. Un sous-comité du Groupe consultatif technique sur les produits de diagnostic des maladies tropicales négligées de l'Organisation mondiale de la Santé a élaboré un profil de produit cible pour un outil visant à détecter une infection par T. b. gambiense. Cet outil doit être suffisamment sensible pour déceler la présence de T. b. gambiense mais suffisamment simple pour être utilisé dans les régions rurales du continent. Idéalement, il doit pouvoir être employé par toute personne ayant reçu une formation sommaire, tant dans des structures de santé périphériques basiques qu'au sein d'une équipe mobile dans un village doté d'infrastructures restreintes. Par ailleurs, il doit fonctionner dans une atmosphère chaude et humide. La formation requise pour son utilisation doit durer moins de deux heures et le test doit être effectué en moins de cinq étapes, sans exiger d'équipement spécifique ni de manipulation délicate. Cet outil doit fournir un résultat fiable en moins de 20 minutes, facile à interpréter, et un seul test doit suffire à établir un traitement. Enfin, afin d'organiser un dépistage de masse, son coût unitaire ne doit pas dépasser un dollar américain.


La tripanosomiasis humana africana es una infección parasitaria potencialmente mortal endémica del África Subsahariana. Alrededor del 95% de los casos se deben al Trypanosoma brucei gambiense, presente en África Occidental y Central. Los signos y síntomas clínicos no son específicos, las pruebas diagnósticas actuales no son suficientemente precisas y la confirmación parasitológica de la infección requiere el examen microscópico de los fluidos corporales y técnicas especializadas de concentración de parásitos. Además, el tratamiento actual no se recomienda a partir de la sola sospecha porque no es suficientemente seguro. La disponibilidad de una prueba diagnóstica sencilla y precisa para identificar a las personas con parásitos ampliaría el tratamiento y ayudaría a disminuir la prevalencia de la enfermedad. Un subcomité del Grupo de Asesoramiento Técnico sobre Diagnóstico de Enfermedades Tropicales Desatendidas de la Organización Mundial de la Salud ha desarrollado un perfil de producto objetivo para una herramienta de diagnóstico destinada a identificar la infección por T. b. gambiense. Esta herramienta debe tener una alta sensibilidad para detectar T. b. gambiense, pero ser lo suficientemente sencilla para su uso en las regiones rurales de África. Lo ideal sería que la herramienta pudiera ser aplicada por cualquier persona mínimamente capacitada en un centro sanitario periférico poco sofisticado o por un equipo móvil en un pueblo con poca infraestructura. La prueba debería funcionar en condiciones de calor y humedad. La formación básica debe durar menos de 2 horas y la prueba debe constar de menos de cinco pasos. No debe necesitarse instrumentación ni manipulación precisa de líquidos. La prueba debe dar un resultado cualitativo en menos de 20 minutos que pueda observarse fácilmente y debe bastar una prueba para determinar el tratamiento. Su coste unitario, inferior a un dólar estadounidense, permitiría un cribado masivo.


Asunto(s)
Líquidos Corporales , Tripanosomiasis Africana , Animales , Humanos , Trypanosoma brucei gambiense , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/tratamiento farmacológico , Tripanosomiasis Africana/epidemiología , África , Pruebas Diagnósticas de Rutina
6.
PLoS Biol ; 17(1): e3000105, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30633739

RESUMEN

Human African trypanosomiasis (HAT), or African sleeping sickness, is a fatal disease found throughout sub-Saharan Africa. The disease is close to elimination in many areas, although it was similarly close to elimination once before and subsequently reemerged, despite seemingly low rates of transmission. Determining how these foci persisted and overcame an apparent transmission paradox is key to finally eliminating HAT. By assessing clinical, laboratory, and mathematical data, we propose that asymptomatic infections contribute to transmission through the presence of an overlooked reservoir of skin-dwelling parasites. Our assessment suggests that a combination of asymptomatic and parasitaemic cases is sufficient to maintain transmission at foci without animal reservoirs, and we argue that the current policy not to treat asymptomatic HAT should be reconsidered.


Asunto(s)
Tripanosomiasis Africana/etiología , Tripanosomiasis Africana/transmisión , África del Sur del Sahara/epidemiología , Animales , Infecciones Asintomáticas , Portador Sano/metabolismo , Humanos , Enfermedades Desatendidas/terapia , Tripanosomiasis Africana/tratamiento farmacológico
7.
Clin Infect Dis ; 73(1): 12-20, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32638003

RESUMEN

BACKGROUND: The diagnosis of gambiense human African trypanosomiasis (gHAT) typically involves 2 steps: a serological screen, followed by the detection of living trypanosome parasites in the blood or lymph node aspirate. Live parasites can, however, remain undetected in some seropositive individuals, who, we hypothesize, are infected with Trypanosoma brucei gambiense parasites in their extravascular dermis. METHODS: To test this hypothesis, we conducted a prospective observational cohort study in the gHAT focus of Forecariah, Republic of Guinea. Of the 5417 subjects serologically screened for gHAT, 66 were enrolled into our study and underwent a dermatological examination. At enrollment, 11 seronegative, 8 unconfirmed seropositive, and 18 confirmed seropositive individuals had blood samples and skin biopsies taken and examined for trypanosomes by molecular and immunohistological methods. RESULTS: In seropositive individuals, dermatological symptoms were significantly more frequent, relative to seronegative controls. T.b. gambiense parasites were present in the blood of all confirmed cases (n = 18) but not in unconfirmed seropositive individuals (n = 8). However, T. brucei parasites were detected in the extravascular dermis of all unconfirmed seropositive individuals and all confirmed cases. Skin biopsies of all treated cases and most seropositive untreated individuals progressively became negative for trypanosomes 6 and 20 months later. CONCLUSIONS: Our results highlight the skin as a potential reservoir for African trypanosomes, with implications for our understanding of this disease's epidemiology in the context of its planned elimination and underlining the skin as a novel target for gHAT diagnostics.


Asunto(s)
Tripanosomiasis Africana , Animales , Guinea , Humanos , Estudios Prospectivos , Trypanosoma brucei gambiense , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/epidemiología
8.
Exp Parasitol ; 219: 108014, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33011238

RESUMEN

The objective set by WHO to reach elimination of human African trypanosomiasis (HAT) as a public health problem by 2020 is being achieved. The next target is the interruption of gambiense-HAT transmission in humans by 2030. To monitor progress towards this target, in areas where specialized local HAT control capacities will disappear, is a major challenge. Test specimens should be easily collectable and safely transportable such as dried blood spots (DBS). Monitoring tests performed in regional reference centres should be reliable, cheap and allow analysis of large numbers of specimens in a high-throughput format. The aim of this study was to assess the analytical sensitivity of Loopamp, M18S quantitative real-time PCR (M18S qPCR) and TgsGP qPCR as molecular diagnostic tests for the presence of Trypanosoma brucei gambiense in DBS. The sensitivity of the Loopamp test, with a detection limit of 100 trypanosomes/mL, was in the range of parasitaemias commonly observed in HAT patients, while detection limits for M18S and TgsGP qPCR were respectively 1000 and 10,000 trypanosomes/mL. None of the tests was entirely suitable for high-throughput use and further development and implementation of sensitive high-throughput molecular tools for monitoring HAT elimination are needed.


Asunto(s)
Técnicas de Diagnóstico Molecular/normas , Técnicas de Amplificación de Ácido Nucleico/normas , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Trypanosoma brucei gambiense/aislamiento & purificación , Tripanosomiasis Africana/prevención & control , Algoritmos , Animales , Recolección de Muestras de Sangre/métodos , Recolección de Muestras de Sangre/normas , ADN Protozoario/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento/métodos , Ensayos Analíticos de Alto Rendimiento/normas , Humanos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Sensibilidad y Especificidad , Manejo de Especímenes/métodos , Manejo de Especímenes/normas , Trypanosoma brucei gambiense/genética , Tripanosomiasis Africana/sangre , Tripanosomiasis Africana/diagnóstico
9.
Lancet ; 390(10110): 2397-2409, 2017 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-28673422

RESUMEN

Human African trypanosomiasis (sleeping sickness) is a parasitic infection that almost invariably progresses to death unless treated. Human African trypanosomiasis caused devastating epidemics during the 20th century. Thanks to sustained and coordinated efforts over the past 15 years, the number of reported cases has fallen to an historically low level. Fewer than 3000 cases were reported in 2015, and the disease is targeted for elimination by WHO. Despite these recent successes, the disease is still endemic in parts of sub-Saharan Africa, where it is a considerable burden on rural communities, most notably in central Africa. Since patients are also reported from non-endemic countries, human African trypanosomiasis should be considered in differential diagnosis for travellers, tourists, migrants, and expatriates who have visited or lived in endemic areas. In the absence of a vaccine, disease control relies on case detection and treatment, and vector control. Available drugs are suboptimal, but ongoing clinical trials provide hope for safer and simpler treatments.


Asunto(s)
Enfermedades Endémicas/estadística & datos numéricos , Salud Global , Enfermedades Desatendidas/epidemiología , Trypanosoma brucei gambiense/aislamiento & purificación , Tripanosomiasis Africana/tratamiento farmacológico , Tripanosomiasis Africana/epidemiología , África del Sur del Sahara/epidemiología , Animales , Países en Desarrollo , Enfermedades Endémicas/prevención & control , Femenino , Humanos , Incidencia , Masculino , Enfermedades Desatendidas/diagnóstico , Medición de Riesgo , Índice de Severidad de la Enfermedad , Viaje , Tripanosomiasis Africana/diagnóstico , Organización Mundial de la Salud
11.
Clin Infect Dis ; 63(9): 1189-1197, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27470243

RESUMEN

BACKGROUND: Human African trypanosomiasis (HAT) caused by Trypanosoma brucei gambiense can be diagnosed in the early hemolymphatic stage (stage 1 [S1]) or meningoencephalitic stage (stage 2 [S2]). Importantly, individuals harbouring high and specific antibody responses to Tbg antigens but negative parasitology are also diagnosed in the field (seropositive [SERO]). Whereas some develop the disease in the months following their initial diagnosis (SERO/HAT), others remain parasitologically negative for long periods (SERO) and are apparently able to control infection. Human leucocyte antigen (HLA)-G, an immunosuppressive molecule, could play a critical role in this variability of progression between infection and disease. METHODS: Soluble HLA-G (sHLA-G) was measured in plasma for patients in the SERO (n = 65), SERO/HAT (n = 14), or HAT (n = 268) group and in cerebrospinal fluid for patients in S1 (n = 55), early S2 (n = 93), or late S2 (n = 110). Associations between these different statuses and the soluble level or genetic polymorphisms of HLA-G were explored. RESULTS: Plasma sHLA-G levels were significantly higher in HAT (P = 6 × 10-7) and SERO/HAT (P = .007) than SERO patients. No difference was observed between the SERO/HAT and HAT groups. Within the HAT group, specific haplotypes (HG010102 and HG0103) displayed increased frequencies in S1 (P = .013) and late S2 (P = .036), respectively. CONCLUSIONS: These results strongly suggest the involvement of HLA-G in HAT disease progression. Importantly, high plasma sHLA-G levels in SERO patients could be predictive of subsequent disease development and could represent a serological marker to help guide therapeutic decision making. Further studies are necessary to assess the predictive nature of HLA-G and to estimate both sensitivity and specificity.


Asunto(s)
Antígenos HLA-G/sangre , Tripanosomiasis Africana/sangre , Adulto , Biomarcadores/sangre , Progresión de la Enfermedad , Femenino , Haplotipos , Humanos , Masculino , Pronóstico , Trypanosoma brucei gambiense , Tripanosomiasis Africana/fisiopatología , Tripanosomiasis Africana/prevención & control
12.
PLoS Pathog ; 10(11): e1004469, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375156

RESUMEN

In West Africa, Trypanosoma brucei gambiense, causing human African trypanosomiasis (HAT), is associated with a great diversity of infection outcomes. In addition to patients who can be diagnosed in the early hemolymphatic phase (stage 1) or meningoencephalitic phase (stage 2), a number of individuals can mount long-lasting specific serological responses while the results of microscopic investigations are negative (SERO TL+). Evidence is now increasing to indicate that these are asymptomatic subjects with low-grade parasitemia. The goal of our study was to investigate the type of immune response occurring in these "trypanotolerant" subjects. Cytokines levels were measured in healthy endemic controls (n = 40), stage 1 (n = 10), early stage 2 (n = 19), and late stage 2 patients (n = 23) and in a cohort of SERO TL+ individuals (n = 60) who were followed up for two years to assess the evolution of their parasitological and serological status. In contrast to HAT patients which T-cell responses appeared to be activated with increased levels of IL2, IL4, and IL10, SERO TL+ exhibited high levels of proinflammatory cytokines (IL6, IL8 and TNFα) and an almost absence of IL12p70. In SERO TL+, high levels of IL10 and low levels of TNFα were associated with an increased risk of developing HAT whereas high levels of IL8 predicted that serology would become negative. Further studies using high throughput technologies, hopefully will provide a more detailed view of the critical molecules or pathways underlying the trypanotolerant phenotype.


Asunto(s)
Inmunidad Innata , Interleucina-10/inmunología , Interleucina-8/inmunología , Trypanosoma brucei gambiense/inmunología , Tripanosomiasis Africana/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Interleucina-10/sangre , Interleucina-8/sangre , Masculino , Persona de Mediana Edad , Trypanosoma brucei gambiense/metabolismo , Tripanosomiasis Africana/sangre , Factor de Necrosis Tumoral alfa/sangre
13.
J Infect Dis ; 212(12): 1996-8, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26080371

RESUMEN

To assess the efficacy of treatment for human African trypanosomiasis, accurate tests that can discriminate relapse from cure are needed. We report the first data that the spliced leader (SL) RNA is a more specific marker for cure of human African trypanosomiasis than parasite DNA. In blood samples obtained from 61 patients in whom human African trypanosomiasis was cured, SL RNA detection had specificities of 98.4%-100%, while DNA detection had a specificity of only 77%. Data from our proof-of-concept study show that SL RNA detection has high potential as a test of cure.


Asunto(s)
ADN Protozoario/análisis , Monitoreo de Drogas/métodos , ARN Lider Empalmado/análisis , Trypanosoma brucei gambiense/aislamiento & purificación , Tripanosomiasis Africana/tratamiento farmacológico , ADN Protozoario/genética , Humanos , ARN Lider Empalmado/genética , Sensibilidad y Especificidad , Trypanosoma brucei gambiense/genética
14.
Trop Med Int Health ; 19(7): 828-31, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24750463

RESUMEN

OBJECTIVES: The immune trypanolysis test (TL) is an accurate sero-diagnostic tool increasingly implemented for sleeping sickness medical surveillance, but it is restricted to the reference laboratories. To facilitate storage and transport of the test specimen, we developed a protocol for the examination of blood spotted on filter paper (TL-fp) that can be stored and shipped at ambient temperature. We compared its performance with the classical TL on plasma (TL-pl) that needs to be kept frozen until use. METHODS: The study was conducted in active foci of the Republic of Guinea. In total, 438 specimens from treated and untreated sleeping sickness patients and serological suspects were tested with both methods. RESULT: TL-fp gave significantly less positive results than TL-pl, but all the confirmed sleeping sickness cases were positive with the TL-fp protocol. CONCLUSION: TL-fp appears to offer a good compromise between feasibility and sensitivity to detect currently infected subjects who play a role in the transmission of Trypanosoma brucei gambiense and is useful for contributing to the elimination of gambiense sleeping sickness.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Vigilancia de la Población/métodos , Trypanosoma brucei gambiense/inmunología , Tripanosomiasis Africana/epidemiología , Animales , Guinea/epidemiología , Humanos , Tamizaje Masivo/métodos , Enfermedades Desatendidas/epidemiología , Sensibilidad y Especificidad , Manejo de Especímenes/métodos , Tripanosomiasis Africana/sangre , Tripanosomiasis Africana/diagnóstico
15.
Parasite ; 31: 15, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38520091

RESUMEN

Trypanosoma brucei gambiense (Tbg) group 2 is a subgroup of trypanosomes able to infect humans and is found in West and Central Africa. Unlike other agents causing sleeping sickness, such as Tbg group 1 and Trypanosoma brucei rhodesiense, Tbg2 lacks the typical molecular markers associated with resistance to human serum. Only 36 strains of Tbg2 have been documented, and therefore, very limited research has been conducted despite their zoonotic nature. Some of these strains are only available in their procyclic form, which hinders human serum resistance assays and mechanistic studies. Furthermore, the understanding of Tbg2's potential to infect tsetse flies and mammalian hosts is limited. In this study, 165 Glossina palpalis gambiensis flies were experimentally infected with procyclic Tbg2 parasites. It was found that 35 days post-infection, 43 flies out of the 80 still alive were found to be Tbg2 PCR-positive in the saliva. These flies were able to infect 3 out of the 4 mice used for blood-feeding. Dissection revealed that only six flies in fact carried mature infections in their midguts and salivary glands. Importantly, a single fly with a mature infection was sufficient to infect a mammalian host. This Tbg2 transmission success confirms that Tbg2 strains can establish in tsetse flies and infect mammalian hosts. This study describes an effective in vivo protocol for transforming Tbg2 from procyclic to bloodstream form, reproducing the complete Tbg2 cycle from G. p. gambiensis to mice. These findings provide valuable insights into Tbg2's host infectivity, and will facilitate further research on mechanisms of human serum resistance.


Title: Cycle de vie expérimental in vivo de Trypanosoma brucei gambiense groupe 2 : de la forme procyclique à la forme sanguicole. Abstract: Trypanosoma brucei gambiense (Tbg) groupe 2 est un sous-groupe de trypanosomes capables d'infecter l'Homme, présent en Afrique de l'Ouest et en Afrique centrale. Contrairement aux autres agents responsables de la maladie du sommeil, tels que Tbg groupe 1 et Trypanosoma brucei rhodesiense, Tbg2 ne présente pas les marqueurs moléculaires habituellement associés à la résistance au sérum humain. Seules trente-six souches de Tbg2 ont été répertoriées, limitant considérablement les recherches sur ce sous-groupe malgré sa nature zoonotique. Certaines de ces souches ne sont disponibles que sous leur forme procyclique, ce qui freine la réalisation des tests de résistance au sérum humain et les études mécanistiques. De plus, la compréhension du potentiel de Tbg2 à infecter les glossines et les hôtes mammifères est limitée. Dans cette étude, 165 glossines Glossina palpalis gambiensis ont été infectées expérimentalement par des parasites Tbg2 sous leur forme procyclique. Trente-cinq jours après l'infection, 43 des 80 glossines encore en vie se sont révélées positives à Tbg2 en PCR sur leur salive. Ces glossines ont réussi à infecter trois des quatre souris utilisées pour leur repas de sang. La dissection des glossines a révélé que seules six d'entre elles étaient réellement porteuses d'infections matures dans leur intestin et leurs glandes salivaires. Il est important de noter qu'une seule glossine porteuse d'une infection mature a suffi pour infecter un hôte mammifère. Ce succès de transmission de Tbg2 confirme que les souches de Tbg2 peuvent s'établir dans les glossines et infecter des hôtes mammifères. Cette étude décrit un protocole in vivo pour transformer la forme procyclique de Tbg2 en forme sanguicole, en reproduisant le cycle complet de Tbg2 de G. p. gambiensis à la souris. Ces résultats fournissent des informations précieuses sur le potentiel infectieux de Tbg2 et faciliteront la recherche sur les mécanismes de résistance au sérum humain des souches.


Asunto(s)
Trypanosoma brucei brucei , Trypanosoma , Tripanosomiasis Africana , Moscas Tse-Tse , Animales , Humanos , Ratones , Trypanosoma brucei gambiense , Tripanosomiasis Africana/parasitología , Moscas Tse-Tse/parasitología , Estadios del Ciclo de Vida , Mamíferos
16.
Infect Dis Poverty ; 13(1): 53, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38978124

RESUMEN

BACKGROUND: Serological screening tests play a crucial role to diagnose gambiense human African trypanosomiasis (gHAT). Presently, they preselect individuals for microscopic confirmation, but in future "screen and treat" strategies they will identify individuals for treatment. Variability in reported specificities, the development of new rapid diagnostic tests (RDT) and the hypothesis that malaria infection may decrease RDT specificity led us to evaluate the specificity of 5 gHAT screening tests. METHODS: During active screening, venous blood samples from 1095 individuals from Côte d'Ivoire and Guinea were tested consecutively with commercial (CATT, HAT Sero-K-SeT, Abbott Bioline HAT 2.0) and prototype (DCN HAT RDT, HAT Sero-K-SeT 2.0) gHAT screening tests and with a malaria RDT. Individuals with ≥ 1 positive gHAT screening test underwent microscopy and further immunological (trypanolysis with T.b. gambiense LiTat 1.3, 1.5 and 1.6; indirect ELISA/T.b. gambiense; T.b. gambiense inhibition ELISA with T.b. gambiense LiTat 1.3 and 1.5 VSG) and molecular reference laboratory tests (PCR TBRN3, 18S and TgsGP; SHERLOCK 18S Tids, 7SL Zoon, and TgsGP; Trypanozoon S2-RT-qPCR 18S2, 177T, GPI-PLC and TgsGP in multiplex; RT-qPCR DT8, DT9 and TgsGP in multiplex). Microscopic trypanosome detection confirmed gHAT, while other individuals were considered gHAT free. Differences in fractions between groups were assessed by Chi square and differences in specificity between 2 tests on the same individuals by McNemar. RESULTS: One gHAT case was diagnosed. Overall test specificities (n = 1094) were: CATT 98.9% (95% CI: 98.1-99.4%); HAT Sero-K-SeT 86.7% (95% CI: 84.5-88.5%); Bioline HAT 2.0 82.1% (95% CI: 79.7-84.2%); DCN HAT RDT 78.2% (95% CI: 75.7-80.6%); and HAT Sero-K-SeT 2.0 78.4% (95% CI: 75.9-80.8%). In malaria positives, gHAT screening tests appeared less specific, but the difference was significant only in Guinea for Abbott Bioline HAT 2.0 (P = 0.03) and HAT Sero-K-Set 2.0 (P = 0.0006). The specificities of immunological and molecular laboratory tests in gHAT seropositives were 98.7-100% (n = 399) and 93.0-100% (n = 302), respectively. Among 44 reference laboratory test positives, only the confirmed gHAT patient and one screening test seropositive combined immunological and molecular reference laboratory test positivity. CONCLUSIONS: Although a minor effect of malaria cannot be excluded, gHAT RDT specificities are far below the 95% minimal specificity stipulated by the WHO target product profile for a simple diagnostic tool to identify individuals eligible for treatment. Unless specificity is improved, an RDT-based "screen and treat" strategy would result in massive overtreatment. In view of their inconsistent results, additional comparative evaluations of the diagnostic performance of reference laboratory tests are indicated for better identifying, among screening test positives, those at increased suspicion for gHAT. TRIAL REGISTRATION: The trial was retrospectively registered under NCT05466630 in clinicaltrials.gov on July 15 2022.


Asunto(s)
Sensibilidad y Especificidad , Trypanosoma brucei gambiense , Tripanosomiasis Africana , Humanos , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/sangre , Côte d'Ivoire , Trypanosoma brucei gambiense/inmunología , Trypanosoma brucei gambiense/aislamiento & purificación , Adulto , Guinea , Estudios Prospectivos , Masculino , Adolescente , Femenino , Adulto Joven , Persona de Mediana Edad , Pruebas Serológicas/métodos , Niño , Ensayo de Inmunoadsorción Enzimática/métodos , Anciano , Preescolar , Anticuerpos Antiprotozoarios/sangre
17.
Med Trop Sante Int ; 3(1)2023 03 31.
Artículo en Francés | MEDLINE | ID: mdl-37525637

RESUMEN

Human African Trypanosomiasis (HAT) is caused by Trypanosoma brucei which is transmitted by the tsetse fly insect vector (Glossina spp). It is one of the 20 Neglected Tropical Diseases (NTD) listed by the WHO. These diseases affect the poorest and most vulnerable communities, for which the WHO has established a dedicated 2021-2030 roadmap. At the time of Alphonse Laveran, HAT devastated the African continent. In the 1960s, the disease was nearly under control, but it strongly re-emerged in the 1990s. A coordinated effort of all stakeholders, with national control programs as the main actors, a strong contribution of research and important donations by the private sector, allowed to decrease the HAT burden significantly. Since 2018, less than 1000 cases are detected annually. We here review new diagnostics, treatments and vector control tools that have been implemented jointly and successfully in several endemic countries.The next key challenge will be to sustain the gains. Newly emerging research questions include long-term carriage of trypanosomes and adaptation of tools to low prevalence contexts. Challenges out of the research area comprise the continued need of funding, maintenance of dedicated human resources, and the key question of access. Sustainable elimination as "interruption of transmission", which is the 2030 NTD roadmap target, can be reached, if these challenges are solved. We stress the importance of continuing to combine the efforts in the fight against the disease, because sustainable elimination of HAT is the best long-term prevention strategy against re-emergence. As such, HAT elimination can serve as an example for other infectious diseases.


Asunto(s)
Trypanosoma brucei brucei , Tripanosomiasis Africana , Moscas Tse-Tse , Animales , Humanos , Tripanosomiasis Africana/epidemiología , Trypanosoma brucei gambiense , Insectos Vectores , Enfermedades Desatendidas/epidemiología
18.
PLoS Negl Trop Dis ; 17(7): e0011528, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37498955

RESUMEN

Although studies on African Trypanosomiases revealed a variety of trypanosome species in the blood of various animal taxa, animal reservoirs of Trypanosoma brucei gambiense and anatomical niches such as skin have been overlooked in most epidemiological settings. This study aims to update epidemiological data on trypanosome infections in animals from human African trypanosomiasis (HAT) foci of Cameroon. Blood and skin snips were collected from 291 domestic and wild animals. DNA was extracted from blood and skin snips and molecular approaches were used to identify different trypanosomes species. Immunohistochemical analyses were used to confirm trypanosome infections in skin snips. PCR revealed 137 animals (47.1%) with at least one trypanosome species in the blood and/or in the skin. Of these 137 animals, 90 (65.7%) and 32 (23.4%) had trypanosome infections respectively in the blood and skin. Fifteen (10.9%) animals had trypanosome infections in both blood and skin snip. Animals from the Campo HAT focus (55.0%) were significantly (X2 = 17.6; P< 0.0001) more infected than those (29.7%) from Bipindi. Trypanosomes of the subgenus Trypanozoon were present in 27.8% of animals while T. vivax, T. congolense forest type and savannah type were detected in 16.5%, 10.3% and 1.4% of animals respectively. Trypanosoma b. gambiense infections were detected in the blood of 7.6% (22/291) of animals. No T. b. gambiense infection was detected in skin. This study highlights the presence of several trypanosome species in the blood and skin of various wild and domestic animals. Skin appeared as an anatomical reservoir for trypanosomes in animals. Despite methodological limitations, pigs, sheep, goats and wild animals were confirmed as potential reservoirs of T. b. gambiense. These animal reservoirs must be considered for the designing of control strategies that will lead to sustainable elimination of HAT.


Asunto(s)
Trypanosoma , Tripanosomiasis Africana , Moscas Tse-Tse , Humanos , Animales , Porcinos , Ovinos , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/veterinaria , Camerún/epidemiología , Prevalencia , ADN Protozoario/genética , ADN Protozoario/química , Trypanosoma/genética , Trypanosoma brucei gambiense/genética , Animales Salvajes , Cabras , Moscas Tse-Tse/genética
19.
PLoS Negl Trop Dis ; 17(11): e0011730, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37943881

RESUMEN

Animal African trypanosomosis is an important vector-borne disease of livestock in sub-Saharan Africa. Pigs seem relatively tolerant to trypanosome infection and could act as a reservoir of trypanosomes affecting animals and humans. Our ability to reliably detect trypanosome infection in pigs depends on the performance of diagnostic tools, which is not well known. In pigs experimentally infected with Trypanosoma brucei brucei, we evaluated the performance of parasitological Buffy Coat Technique (BCT), two molecular (TBR and 5.8S PCR) and four serological tests (CATT, HAT Sero-K-Set rapid diagnostic test-RDT, indirect ELISA, immune trypanolysis). Most diagnostic tests showed high specificity, estimated at 100% (95% CI = 74-100%) with the exception of CATT and RDT whose specificity varied between 100% (95% CI = 74-100%) to 50% (95% CI = 7-93%) during the experiment. The sensitivity of each test fluctuated over the course of the infection. The percentage of positive BCT over the infection (30%) was lower than of positive PCR (56% and 62%, depending on primers). Among the serological tests, the percentage of positive tests was 97%, 96%, 86% and 84% for RDT, ELISA, immune trypanolysis and CATT, respectively. Fair agreement was observed between both molecular tests (κ = 0.36). Among the serological tests, the agreement between the ELISA and the RDT was substantial (κ = 0.65). Our results on the T.b. brucei infection model suggest that serological techniques are efficient in detecting the chronic phase of infection, PCR is able to detect positive samples several months after parasites inoculation while BCT becomes negative. BCT examination and RDT are useful to get a quick information in the field, and BCT can be used for treatment decision. ELISA appears most suited for epidemiological studies. The selection of diagnostic tests for trypanosomosis in pigs depends on the context, the objectives and the available resources.


Asunto(s)
Trypanosoma brucei brucei , Trypanosoma , Tripanosomiasis Africana , Humanos , Animales , Porcinos , Tripanosomiasis Africana/diagnóstico , Tripanosomiasis Africana/veterinaria , Tripanosomiasis Africana/parasitología , Ganado , Pruebas Diagnósticas de Rutina , Sensibilidad y Especificidad
20.
Parasite ; 30: 36, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37728508

RESUMEN

African trypanosomoses, whose pathogens are transmitted by tsetse flies, are a threat to animal and human health. Tsetse flies observed at the military base of the French Forces in Côte d'Ivoire (FFCI base) were probably involved in the infection and death of military working dogs. Entomological and parasitological surveys were carried out during the rainy and dry seasons using "Vavoua" traps to identify tsetse fly species, their distribution, favorable biotopes and food sources, as well as the trypanosomes they harbor. A total of 1185 Glossina palpalis palpalis tsetse flies were caught, corresponding to a high average apparent density of 2.26 tsetse/trap/day. The results showed a heterogeneous distribution of tsetse at the FFCI base, linked to more or less favorable biotopes. No significant variation in tsetse densities was observed according to the season. The overall trypanosomes infection rate according to microscopic observation was 13.5%. Polymerase chain reaction (PCR) analyses confirmed the presence of Trypanosoma vivax and T. congolense forest type, responsible for African animal trypanosomosis. Our findings suggest that there is a risk of introduction and transmission of T. brucei gambiense, responsible for human African trypanosomiasis, on the study site. This risk of transmission of African trypanosomes concerns not only the FFCI base, but also inhabited peripheral areas. Our study confirmed the need for vector control adapted to the eco-epidemiological context of the FFCI base.


Title: Écologie des mouches tsé-tsé et risque de transmission des trypanosomes africains lié à une zone forestière protégée dans une base militaire de la ville d'Abidjan, Côte d'Ivoire. Abstract: Les trypanosomoses africaines, dont les agents pathogènes sont transmis par les mouches tsé-tsé, constituent une contrainte pour la santé animale et humaine. Des mouches tsé-tsé observées dans la base militaire des Forces françaises en Côte d'Ivoire (base FFCI) ont probablement été impliquées dans l'infection et la mort de chiens militaires. Des enquêtes entomologiques et parasitologiques ont été menées pendant la saison pluvieuse et la saison sèche à l'aide de pièges "Vavoua" afin d'identifier les espèces de mouches tsé-tsé, leur distribution, les biotopes favorables et leur source de nourriture ainsi que les trypanosomes qu'elles hébergent. Au total 1185 mouches tsé-tsé de l'espèce Glossina palpalis palpalis ont été capturées, ce qui correspond à une densité apparente moyenne élevée de 2,26 tsé-tsé/piège/jour. Les résultats ont montré une distribution hétérogène des tsé-tsé dans la base FFCI en lien avec des biotopes plus ou moins favorables. Aucune variation significative des densités de tsé-tsé n'a été observée en fonction de la saison. Le taux d'infection global par les trypanosomes était de 13,5 % selon l'observation microscopique. Les analyses PCR ont confirmé la présence de Trypanosoma vivax et T. congolense type forêt, responsable de la trypanosomose animale africaine. Nos résultats suggèrent qu'il existe un risque potentiel d'introduction et de transmission de T. brucei gambiense responsable de la trypanosomiase humaine africaine dans la zone d'étude. Ce risque de transmission des trypanosomes africains concerne non seulement l'intérieur de la base FFCI, mais aussi les espaces périphériques habités. Notre étude a confirmé la nécessité de mener une lutte antivectorielle adaptée au contexte éco-épidémiologique de la base FFCI.


Asunto(s)
Trypanosoma , Tripanosomiasis Africana , Moscas Tse-Tse , Animales , Perros , Humanos , Côte d'Ivoire/epidemiología , Instalaciones Militares , Tripanosomiasis Africana/epidemiología , Tripanosomiasis Africana/veterinaria , Bosques
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA